WO2015044762A1 - 7-aminocephalosporanic acid derivative as inhibitor of il-15 and il-2 activity - Google Patents

7-aminocephalosporanic acid derivative as inhibitor of il-15 and il-2 activity Download PDF

Info

Publication number
WO2015044762A1
WO2015044762A1 PCT/IB2014/001940 IB2014001940W WO2015044762A1 WO 2015044762 A1 WO2015044762 A1 WO 2015044762A1 IB 2014001940 W IB2014001940 W IB 2014001940W WO 2015044762 A1 WO2015044762 A1 WO 2015044762A1
Authority
WO
WIPO (PCT)
Prior art keywords
cefazolin
cells
treatment
pbmc
activity
Prior art date
Application number
PCT/IB2014/001940
Other languages
French (fr)
Inventor
Katarzyna KOZIAK
Barbara ŻYŻYŃSKA-GRANICA
Sławomir FILIPEK
Szymon NIEWIECZERZAŁ
Bartosz TRZASKOWSKI
Oliwia Zegrocka-Stendel
Małgorzata DUTKIEWICZ
Piotr KRZECZYŃSKI
Elżbieta KACZMAREK
Magdalena Winiarska
Original Assignee
Warszawski Uniwersytet Medyczny
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Warszawski Uniwersytet Medyczny filed Critical Warszawski Uniwersytet Medyczny
Priority to EP14799527.8A priority Critical patent/EP3068491B1/en
Priority to PL417879A priority patent/PL235635B1/en
Priority to PL14799527T priority patent/PL3068491T3/en
Priority to CA2925652A priority patent/CA2925652A1/en
Priority to AU2014326355A priority patent/AU2014326355B2/en
Priority to US15/026,125 priority patent/US20160235762A1/en
Priority to DK14799527.8T priority patent/DK3068491T3/en
Priority to ES14799527T priority patent/ES2747635T3/en
Publication of WO2015044762A1 publication Critical patent/WO2015044762A1/en
Priority to US15/661,586 priority patent/US11452728B2/en
Priority to AU2019236714A priority patent/AU2019236714B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/542Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/545Compounds containing 5-thia-1-azabicyclo [4.2.0] octane ring systems, i.e. compounds containing a ring system of the formula:, e.g. cephalosporins, cefaclor, or cephalexine
    • A61K31/546Compounds containing 5-thia-1-azabicyclo [4.2.0] octane ring systems, i.e. compounds containing a ring system of the formula:, e.g. cephalosporins, cefaclor, or cephalexine containing further heterocyclic rings, e.g. cephalothin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection

Definitions

  • Interleukin 15 is the cytokine exerting pleiotropic activity towards immune system cells as well as other cell types. I L-15 exhibits broad spectrum bioactivity, therefore it is placed at the top of the pro-inflammatory cytokines cascade. The impairment of the mechanisms regulating the expression of IL-15 results in the overproduction of this cytokine and contributes directly to the development of such pathologies as inflammatory processes, autoimmune diseases, infections and neoplastic changes. IL-15 is considered a crucial cytokine in the etiology of rheumatoid arthritis (Mclnnes I.B. et al. , Nat. Med. 2, 175-82 (1996; Mclnnes ' l. B.
  • rituximab chimeric monoclonal antibody against the protein CQ20 primarily found on the surface of mature B cells acting by eliminating B cells.
  • Tocilizumab humanized monoclonal antibody against interleukin 6 receptor, has been approved for the European pharmaceutical market as the first IL-6 inhibitor.
  • IL-2 is released predominantly by activated T helper lymphocytes (CD4+), it is also secreted by CD8+ T lymphocytes, dendritic cells and thymus-derived lymphocites (128).
  • the downstream effect of IL-2 occurs through its high-affinity binding to the receptor IL-2R, consisting of three subunits: IL-2Ra subunit, which is IL-2 specific and IL-2RP and IL-2Ry subunits, which are shared with IL-15.
  • Serum soluble form of IL-2 receptor (slL-2Ra, Tac peptide) is released into circulation by the immune cells.
  • the derivatives of 7-aminocephalosporanic acid, cephalosporines are the group of semi-synthetic ⁇ -lactam antibiotics, characterized by broad spectrum of antibacterial activity.
  • the ⁇ -lactam antibiotics exert their effect by interfering with the structural crosslinking of peptidoglycans (murein) in bacterial cell walls. They bind covalently to the active sites of D- alanyl carboxypeptidase and transpeptidase, inhibiting the activity of these bacterial enzymes involved in the synthesis of bacterial cell wall.
  • the specific compound is 7-aminocephalosporanic acid derivative, (6R,7R)-7-[[-2-amino-2- phenylacetyl]amino]-3-methyl-8-oxo-5-thia-1 -azabicyclo[4.2.0]oct-2-en-2-carboxylic acid, known under the International Non-proprietary Name (INN) Cefazolin.
  • the present invention provides 7-aminocephalosporanic acid derivative - Cefazolin, for use as the inhibitor of IL-15 specific IL-15Ra and IL-2RP and IL-2Ry receptor subunits shared by IL- 15 and IL-2, in the prevention and treatment of IL-15 and IL-2 overproduction related diseases.
  • Fig. 1 shows the effect of Cefazolin at different concentration on proliferation of IL-15-stimulated PBMC.
  • Fig. 4 shows the effect of Cefazolin on IL-2-induced TNF-a synthesis in PBMC.
  • Fig. 5 shows the effect of Cefazolin on IL-15-induced IL-17 synthesis in PBMC.
  • Cefazolin is most frequently administered in the treatment of serious infections, such as, for example, respiratory tract infection (in particular lower parts of respiratory tract) due to: S. pneumoniae, group A ⁇ -hemolytic streptococci, Klebsiella spp., H. influenzae, S. aureus; urinary tract and genital infections due to: E. coli, P. mirabilis, Klebsiella spp.; skin and soft tissue infections due to: S. aureus, group A ⁇ -hemolytic streptococci and the other strains of streptococci; bilary tract infections due to E. coli and enterococci, P. mirabili, Klebsiella spp. and S.
  • respiratory tract infection in particular lower parts of respiratory tract
  • S. pneumoniae S. pneumoniae, group A ⁇ -hemolytic streptococci, Klebsiella spp., H. influenzae, S. aureus
  • urinary tract and genital infections due to: E.
  • aureus bone and joint infections due to S. aureus; septicemia due to S. pneumoniae, P. mirabilis, E. coli, Klebsiella spp.; endocarditis due to S. aureus and group A ⁇ -hemolytic streptococci; as well as in perioperative prophylaxis (according to the general indications of the pharmaceutical product).
  • Cefazolin biological activity with respect to the use according to the invention has been proved in vitro.
  • the studies' results confirm that Cefazolin suppresses IL-15-induced cell proliferation and inhibits IL-15-induced synthesis of TNF-a and IL-17.
  • the same inhibitory activity has been observed when cells were stimulated with IL-2.
  • the pharmaceutical composition may be formulated in any pharmaceutical form suitable for the systemic administration, for example oral administration, such as tablets and capsules, starch capsules, coated tablets or enteral tablets; as powders or granules; as a solution, a suspension or an emulsion.
  • Tablets and capsules for oral administration may contain excipients routinely used in pharmaceutical practice, such as binders, diluents, disintegrants or lubricants.
  • the tablets may be coated by any method known in the art.
  • Fig. 3 The effect of Cefazolin at different concentrations (20 ⁇ , 50 ⁇ , 100 ⁇ , 200 ⁇ , 400 ⁇ , 600 ⁇ , 1000 ⁇ ) on IL-15-induced TNF- ⁇ synthesis in PBMC is depicted in Fig. 3.
  • Fig. 4 The effect of Cefazolin at 300 ⁇ concentration on IL-2-induced TNF-a synthesis in PBMC is depicted in Fig. 4.
  • Increased cell proliferation is one of the characteristic responses to IL-15 or IL-2 stimulation.
  • Selective blocking of the IL-15 specific IL-15Ra receptor as well as shared by IL-15 and IL-2, IL-2RP and IL-2Ry the receptor subunits, results in the inhibition of the biological activity of the said cytokines.
  • IL-15 or I L-2-induced increase of cell proliferation does not occur.
  • the effect of Cefazolin on the proliferation of peripheral blood mononuclear cells (PBMC) isolated from blood of healthy donors was evaluated in the assay using a fluorescent CSFE dye (Molecular Dynamics, Great Britain) and in the bromodeoxyuridine incorporation test (BrdU) (BrdU Cell Proliferation Assay, Calbiochem, USA).
  • LDH enzyme is released into the matrix. Increased LDH activity in the culture medium correlates with the increased number of dead cells resulting from the cytotoxic effect of the tested compound. LDH activity measured in cell lysates is used for the assessment of a number of living cells.
  • the results of the following study has demonstrated biological activity of Cefazolin, which inhibits 11-15 or IL-2-induced PBMC proliferation and does not trigger the apoptotic pathway. The experiments were repeated three time on PBMC from different donors.

Abstract

The invention relates to 7-zminocephalosporanic acid derivative, Cefazolin, for use as the inhibitor of IL-15 and IL-2 receptors in the prevention and treatment of IL-15 and IL-2 overproduction related diseases.

Description

7-Aminocephalosporanic acid derivative as inhibitor of IL-15 and IL-2 activity
Field of the invention
The present invention relates to 7-aminocephalosporanic acid derivative for the use in the prevention and treatment of the diseases related to the overproduction of interleukin 15 and interleukin 2, such as, rheumatoid arthritis, psoriasis, inflammatory bowel disease, sarcoidosis, T-cell leukemias or transplant rejection.
Background of the invention
Interleukin 15 (IL-15) is the cytokine exerting pleiotropic activity towards immune system cells as well as other cell types. I L-15 exhibits broad spectrum bioactivity, therefore it is placed at the top of the pro-inflammatory cytokines cascade. The impairment of the mechanisms regulating the expression of IL-15 results in the overproduction of this cytokine and contributes directly to the development of such pathologies as inflammatory processes, autoimmune diseases, infections and neoplastic changes. IL-15 is considered a crucial cytokine in the etiology of rheumatoid arthritis (Mclnnes I.B. et al. , Nat. Med. 2, 175-82 (1996; Mclnnes 'l. B. et al., Nat. Med. 3, 189-95 (1007); Mclnnes I.B. et al. , Immunol Today 19, 75-9 (1998), psoriasis (Villadsen L.S. et al., J. Clin. Invest. 1 12, 1571 -80 (2003), inflammatory bowel disease (Kirman I , Nielsen O.H. , Am. J. Gastroenterol 91 , 1789-1794 (1996); Sakai T. et al. , Gastroenterology 1 14, 1237-1243 (1998)), sarcoidosis (Agostini C.T.L. et al. , J. Immunol. 157, 910-8 (1996)) and T-cell leukemias (Dobbeling U. et al., Blood 92, 252-8 (1998)). Much attention is given to IL-15, on account of participation of this cytokine in transplant rejection (Baan C.C. et al. , Transplant Proc. 31 , 2726-8 (1999); Lewis E.C. et al. , Cytokine 34, 106-13 (2006); Shi R. et al. , Transpl. Immunol. 12, 103-8 (2004); Ferrari-Lacraz S. et al., Transplantation 82, 1510-7 (2006); Zheng X.X. et al. , Transplantation 81 , 109-16 (2006)).
Significant involvement of IL-15 in the development of the pathogeneses of the aforementioned afflictions suggests that targeting this cytokine signaling pathway can reduce or eliminate the occurrence of IL-15-related disorders. This strategy has been proved lately by a number of biological assays. Reduction of IL-15-mediated biological responses, using soluble IL-15Ra receptor (Liew F.Y., Mclnnes I. B. , Ann. Rheum. Dis. 61 Supl. 2, ii100-2 (2002); Ruchatz H. et al., J Immunol. 160, 5664-60 (1998); Smith X.G. et al., J. Immunol. 165, 3444-50 (2000); Wei X et al., J. Immunol. 167, 277-82 (2001 )), antibodies inhibiting IL-2/IL-15RB receptor (Morris J.C., Proc. Natl, Acad. Sci. USA 103, 401 -6 (2001 ); Tinubu S.A. et al., J. Immunol. 153, 4330-8 (1994), antibodies inhibiting IL-15 (Villadsen L.S. et al. J. Clin. Invest. 1 12, 1571 -80 (2003) or modified IL-15 molecule of competitive antagonist activity (Ferrari- Lacraz S. et al. , J. Immunol. 173, 5818-26 (2004); Kim Y.S. et al. , J. Immunol. 160, 5742-8 (1998)) always resulted in the alleviation of the disease symptoms. The experimental therapies demonstrated decreased occurrence of collagen-induced rheumatoid arthritis incidence in mice (Ruchatz H. et al. , J Immunol. 160, 5664-60 (1998); Ferrari-Lacraz S. et al. , J. Immunol. 173, 5818-26 (2004); Kim Y.S. et al. , J. Immunol. 160, 5742-8 (1998)) and primates (Liew F.Y. , clnnes I. B. , Ann. Rheum. Dis. 61 Supl. 2, ii100-2 (2002)), attenuation of psoriasis symptoms in the mice model of the disease (Villadsen L.S. et al., J. Clin. Invest. 1 12, 1571 -80 (2003)), reduction of carrageenan-induced inflammation in mice (Wei X et al. , J. Immunol. 167, 277-82 (2001 )) and, also in mice, prolonged survival of heart allotransplants (Smith X.S. et al. , J. Immunol. 165, 3444-50 (2000); Tinubu S.A. et al. , J. Immunol. 153, 4330-8 (1994) and islets of Langerhans (Ferrari-Lacraz S. et al., J. Immunol. 173, 5818-26 (2004)). Currently adopted strategies based on inhibiting IL-15 seem to be effective, but none of them has been approved for clinical use so far. The most advanced and promising, from the medical application point of view, are the trials aimed at the inhibition of IL-15 activity by anti-IL-15 human antibodies (HuMax-IL15, AMG-714) (Baslund B. et al„ Arthritis Rheum 52, 2686-92 (2005)). However, the matter of serious concern is the effect of 'reverse signaling' (Budagian V. et al., J. Biol. Chem. (2004)). It is assumed that the antibody HuMax-IL15 and IL-15 complex can itself induce a cell response, thus limiting the therapeutic effectiveness of this antibody directed toward IL-15 neutralization (Budagian V. et al. Cytokine Growth Factor Rev. 7, 259-80 (2006).
In addition, our unpublished results demonstrate significant angiogenic activity of IL-15. The participation of IL-15 in angiogenesis in vivo has already been reported (Angiolllo A.L. et al. , Biochem. Biophys. Res. Commun. 233, 231 -7 (1997); Kuniyasu H. et al., Pathobiology 69, 86-95 (2001 )), but our recent outcomes have also shown IL-15-induced proliferation and migration of endothelial cells. The identification of another angiogenic factor was not surprising, because its presence had already been detected in rheumatoid arthritis, where the impaired mechanism of angiogenesis is considered to be the main pathogenesis of this illness.
Rheumatoid arthritis (RA) is the most common systemic disease of connective tissue, which affects on average about 1 % of the world's population. Recent estimates have shown higher prevalence in women than in men (3: 1 ). The highest prevalence of RA has been reported in the 30-60 age range. It is assumed, about 30% of people afflicted with RA suffer from severe symptoms, which are the leading causes of disabilities within several years. The average life span of people suffering from RA is about 10 years shorter, according to the statistical data. It is thought that the onset of RA results from the complex combination of many factors, such as genetic predisposition, impaired innate and acquired immune response as well as environmental components. Pharmacological treatment of RA, currently available on the medical market, does not remove the cause of the disease. Non-steroidal anti-inflammatory drugs (NSAIDs) are medications, which help managing chronic pain (analgesic effect) and reduce the inflammation, when used over a period of time. They suppress the symptoms of the disease, but do not stop the progress of the illness.
Among the disease-modifying antirheumatic drugs (DMARDs), the first line treatment in RA is methotrexate. The other synthetic DMARDs are leflunomide, sulfasalazine, hydroxychloroquine, D-penicillamine, gold salts, azathioprine, cyclosporine and cyclophosphamide. However, even in patients responding to the treatment, the disease progresses and diminished medical efficacy is observed after long term treatment.
The other class of DMARDs are the biologic medical products. Among the drugs available on the pharmaceutical market, the inhibitors of tumor necrosis factor (TNF) are to be mentioned:
infliximab (chimeric anti-TNF monoclonal antibody);
etanercept (fusion protein, consisting of the extracellular receptor domain p75 for TNF and the Fc fragment of the human antibody lgG1 );
adalimumab (human anti-TNF monoclonal antibody),
as well as the inhibitors of other proteins:
anakinra, IL-1 receptor antagonist;
abatacept, fusion protein composed of the Fc region of the immunoglobulin lgG1 fused to the extracellular domain of CTLA-4. By binding B7-1 and B7-2 co-stimulating molecules located on the antigen-presenting cells it inhibits the co-stimulatory signal transduction by CD28 on T cells;
rituximab, chimeric monoclonal antibody against the protein CQ20 primarily found on the surface of mature B cells acting by eliminating B cells.
Tocilizumab, humanized monoclonal antibody against interleukin 6 receptor, has been approved for the European pharmaceutical market as the first IL-6 inhibitor.
Positive results of phase II clinical trials carried out by Amgen company with AMG-714
(previously HuMax-IL15) - human monoclonal antibody that targets IL-15, were disclosed in 2004 (Mclnnes, I., et al.). However, to date there is no available information about the phase III clinical trials.
The introduction of the biologic medical products into rheumatoid arthritis therapy is considered big progress in the RA treatment, but these biologies (usually used in a combination with methotrexate of cytostatic and immunosuppressive properties) are only effective in limiting the disease symptoms and delaying joint degradation in about 30% of treated patients.
Due to the limited therapeutic effects and high production costs of currently available biologic medical products, the demand for an efficacious anti-RA drug of new generation still remains valid. Ongoing research is aimed at discovering new molecular mechanisms for targeted therapies.
Peptides of modified sequences mimicking IL-15 (revealed, among others, in WO 2006/029578 and WO 2010/037351) have also been proposed as potential pharmaceuticals in the rheumatoid arthritis treatment. These peptides are supposed to bind to the receptor IL-15Ra subunit, which should result in the inhibition of T cells proliferation, diminished TNF-a induction as well as expression of IL-8 and IL-6. To date the efficacy of these new compounds has not been proved in clinical studies. There are other cytokines, which participate in the pathogenesis of inflammatory diseases, induced by interleukin 15 overproduction.
I the complex network of the immune mediators, interleukin 2 (IL-2) is regarded the pivotal cytokine controlling proliferation and differentiation of the immune cells. IL-2 activates, among others, proliferation and differentiation of T lymphocytes, differentiation of T lymphocytes towards cytotoxic T lymphocytes, growth and differentiation of B lymphocytes, activation and proliferation of NK cell, and macrophages activation. Under physiological conditions IL-2 is not detected in a blood serum. Due to its contribution to the activation and stimulation of the effector function of the immune cells, this cytokine is considered one of the chief inflammatory mediators in the autoimmune diseases. IL-2 is released predominantly by activated T helper lymphocytes (CD4+), it is also secreted by CD8+ T lymphocytes, dendritic cells and thymus-derived lymphocites (128). The downstream effect of IL-2 occurs through its high-affinity binding to the receptor IL-2R, consisting of three subunits: IL-2Ra subunit, which is IL-2 specific and IL-2RP and IL-2Ry subunits, which are shared with IL-15. Serum soluble form of IL-2 receptor (slL-2Ra, Tac peptide) is released into circulation by the immune cells. Under physiological conditions its concentration in a blood serum is at a very low level, but it is significantly elevated in some pathologies, for example in the autoimmune diseases, inflammations, some types of leukemia, and allogenic transplant rejections. It has been observed, the level of slL-2Ra correlates with the RA progression, there is also the evidence it induces the expression of IL-15 (Release of slL-2R alpha from and activation of native human peripheral blood mononuclear cells by recombinant IL-15. Treiber-Held S, Stewart DM, Barraclough HA, Kurman CC, Nelson DL. Clin Immunol Immunopathol. 1996 Jul;80(1):67-75).
Currently, different components of the IL-2/IL-2R complex and/or their antagonists, for example, murine monoclonal antibodies (anti-TAC-M) directed toward the human slL-2Ra receptor, are used in the medical treatment. Due to their strong immunogenicity, humanized monoclonal antibodies (anti-TAC-H) recently have been engineered and introduced into clinical practice. Anti-Tac (daclizumab, Zenapax) in the treatment of leukemia, autoimmune diseases, and in the prevention of allograft rejection: a 25-year personal odyssey, Waldmann TA. J Clin Immunol. 2007 Jan; 27(1 ): 1-18. The monoclonal antibodies prevent IL-2 binding, among others, to T lymphocytes, thus hampering IL-2 mediated cell response involved in the process of allograft rejection. (WWW.drugbank.ca/drugs/BTD00007).
A need to design a new drug based on a small synthetic molecule has motivated the authors of the present invention to alter the concept of the treatment of the IL-15 overproduction , related diseases. This approach relates to the inhibition of the IL-15 biological activity by a compound selectively binding to IL-15Ra receptor. According to the current state of knowledge and our own unpublished results, the strategy, while implemented, should reduce the proinflammatory cascade as well as angiogenesis caused by pro-inflammatory cytokine IL-15. The additive synergism may be the most likely explanation of the mechanism, so far unrecognized, of the inhibition of IL-15 biological activity by the biologicals. The known small chemical molecules of potential application in rheumatoid arthritis, which interfere with the interaction of the receptor IL-15Ra and its ligand (IL-15) are only phenylpyrazole anilide derivatives, disclosed by Ushio H. et al. in Letters in Drug Design Discovery, 5, 292-296 (2008),. The molecule Y-320, belonging to the aforementioned group of compounds of proven high bioavailability and in vitro activity, inhibits IL-15-induced T cells activity.
The aim of the present invention was to select a molecule, which would effectively inhibit the biological activity of both IL-15 and IL-2 by selective binding to IL-15 specific IL-15Ra receptor and concomitant blocking IL-2Rp as well as IL-2Ry receptor subunits shared by these two cytokines.
The derivatives of 7-aminocephalosporanic acid, cephalosporines, are the group of semi-synthetic β-lactam antibiotics, characterized by broad spectrum of antibacterial activity. The β-lactam antibiotics exert their effect by interfering with the structural crosslinking of peptidoglycans (murein) in bacterial cell walls. They bind covalently to the active sites of D- alanyl carboxypeptidase and transpeptidase, inhibiting the activity of these bacterial enzymes involved in the synthesis of bacterial cell wall.
In US 4,891 ,370 the use of cephalosporanic acid derivatives as anti-inflammatory agents, in particular, of high effectiveness in rheumatoid arthritis, has been disclosed. This antiinflammatory activity has been attributed to the inhibition of elastase activity by cephalosporins. The cephalosporin derivatives, disclosed in US 4,891 ,370, are substituted at position 3 with the organic radical or the group typical for cephalosporins and penems, while the active hydrogen of the ester group at position 2 of the 6-membered ring is the organic radical. Summary of the invention
The identification of a chemical compound which will fit in the IL-15Ra receptor was facilitated due to the evaluation of the receptor domain structure, which is of key importance while binding to IL-15 (Wei X. et al., J. Immunol. 167, 277-82 (2001 )). The search for small chemical molecules fitting in the IL-15 specific receptor domain was carried out on the basis of the model of the [IL-15Ra - IL-15] complex structure, elaborated on the basis of the crystal structure of the complex fragment (2Z3Q code of Protein Data Bank). The assignment of receptor binding sites pharmacophores, while considering specific interactions with IL-15, was enabled due to determination of the 3D structure of the [IL-15Ra - IL-15] complex. It was followed by the search of the small molecules data base ZINC, embracing about 20 million chemical compounds, to find the molecules which can meet the structural requirements for pharmacophores. The screening included more than 10 thousand compounds fulfilling the established criteria. The calculations were also carried out in order to theoretically predict the binding affinity and strength of the selected compounds and the receptor active site. Molecular docking of the chosen molecules was performed using GLIDE software. While docking, the IL- 15 binding site of the receptor remained rigid, but the molecule was able to undergo conformational changes due to the modifications of rotating chemical bonds. Additional docking carried out for the pre-selected 500 molecules allowed the induction fitting in of the two interacting partners (protein receptor and small molecule).
Using the above described screening methods, the specific compound has been selected with the efficiency in inhibiting IL-15 and IL-2-induced cell responses In vitro.
The specific compound is 7-aminocephalosporanic acid derivative, (6R,7R)-7-[[-2-amino-2- phenylacetyl]amino]-3-methyl-8-oxo-5-thia-1 -azabicyclo[4.2.0]oct-2-en-2-carboxylic acid, known under the International Non-proprietary Name (INN) Cefazolin.
The present invention provides 7-aminocephalosporanic acid derivative - Cefazolin, for use as the inhibitor of IL-15 specific IL-15Ra and IL-2RP and IL-2Ry receptor subunits shared by IL- 15 and IL-2, in the prevention and treatment of IL-15 and IL-2 overproduction related diseases.
Brief description of the figures Fig. 1 shows the effect of Cefazolin at different concentration on proliferation of IL-15-stimulated PBMC.
Fig. 2 shows the effect of Cefazolin at different concentration on proliferation of IL-2-stimulated PBMC.
Fig. 3 shows the effect of Cefazolin on IL-15-induced TNF-a synthesis in PBMC.
Fig. 4 shows the effect of Cefazolin on IL-2-induced TNF-a synthesis in PBMC.
Fig. 5 shows the effect of Cefazolin on IL-15-induced IL-17 synthesis in PBMC.
Fig. 6 shows the effect of Cefazolin on IL-2-induced IL-17 synthesis in PBMC. Disclosure of the invention
Cefazolin is first-generation cephalosporin of broad spectrum of antimicrobial activity. Potent bactericidal activity against Gram-positive bacteria is the reason for the frequent indications of this antibiotic in the clinical practice. The spectrum of Cefazolin antibacterial activity comprises: staphylococci; streptococci, including Streptococcus pneumoniae (only penicillin susceptible), Haemophilus influenzae, Escherichia coli, Proteus mirabilis, Klebsiella pneumoniae, anaerobic cocci.
Cefazolin is most frequently administered in the treatment of serious infections, such as, for example, respiratory tract infection (in particular lower parts of respiratory tract) due to: S. pneumoniae, group A β-hemolytic streptococci, Klebsiella spp., H. influenzae, S. aureus; urinary tract and genital infections due to: E. coli, P. mirabilis, Klebsiella spp.; skin and soft tissue infections due to: S. aureus, group A β-hemolytic streptococci and the other strains of streptococci; bilary tract infections due to E. coli and enterococci, P. mirabili, Klebsiella spp. and S. aureus; bone and joint infections due to S. aureus; septicemia due to S. pneumoniae, P. mirabilis, E. coli, Klebsiella spp.; endocarditis due to S. aureus and group A β-hemolytic streptococci; as well as in perioperative prophylaxis (according to the general indications of the pharmaceutical product).
Cefazolin is very poorly absorbed from gastroinstestinal tract, therefore it must be administered parenterally, i.m. or i.v. It has good bioavailability into pleural cavity, peritoneal cavity, synovial fluid, bile, bones, and urinary tract, but it does not cross the blood brain barrier. Cefazolin is not metabolized. It is excreted in urine, due to glomerular filtration mechanism (in a lesser degree by tubular secretion), of unchanged structure at 90% of the excreted amount.
Whenever the term 'Cefazolin' is used hereinafter, it refers to (6f?,7R)-7-([-2-amino-2- phenylacetyl]amino)-3-methyl-8-oxo-5-thia-1-azabicyclo[4.2.0]oct-2-en-2-carboxylic acid or pharmaceutically acceptable salts thereof, in particular sodium salt.
Cefazolin biological activity with respect to the use according to the invention has been proved in vitro. The studies' results confirm that Cefazolin suppresses IL-15-induced cell proliferation and inhibits IL-15-induced synthesis of TNF-a and IL-17. The same inhibitory activity has been observed when cells were stimulated with IL-2.
These findings indicate Cefazolin may be used to hamper excessive cells' responses induced by IL-15 and IL-2, due to blocking IL-15 specific IL-15Ra receptor and IL-2R and IL- 2Rv receptor subunits, shared by these two cytokines.
In particular, Cefazolin may be used in the prevention and treatment of the diseases from the group comprising rheumatoid arthritis, psoriasis, inflammatory bowel disease, sarcoidosis, T-cell leukemias and transplant rejection.
Preferably, Cefazolin may be used in the treatment of rheumatoid arthritis.
According to the invention, Cefazolin can be administered to an individual in the need of such a treatment, in particular to a human, at therapeutically effective dose.
The term 'treatment" refers to the suppression of a state, a disorder or a disease, which means inhibition, reduction or delay of the disease development, recurrence of the illness or at least one of its symptoms, or recovery, meaning the regression of a state, a disorder or a disease or at least one of its symptoms.
The term 'therapeutically effective dose' refers to the amount of the compound sufficient to produce a therapeutic response when administered to an individual in order to cure a state, a disorder or a disease. The 'therapeutically effective dose' will vary depending on the nature of the selected compound and the route of administration, kind of a disease and its status, age, body weight, physical condition, susceptibility to the treatment, and it can be recommended by a medical doctor on the basis of their own experience and the results of clinical trials.
The therapeutic dose of Cefazolin can be administered as a single dose or divided doses given in certain intervals of time, for example as two, three, four or more daily doses. Maximum daily dose of Cefazolin is 12 g.
The daily dose of Cefazolin in adult may be in the range of 250 to 500 mg or it may be even higher, as administered every 8 or 12 h. Although the administration of Cefazolin per se may be considered, in general it will be used as active ingredient of the pharmaceutical formulation, of appropriate form suited to the particular route of administration.
Another embodiment of the invention relates to the use of Cefazolin for manufacturing of the pharmaceutical formulation for the prevention or treatment of the diseases such as rheumatoid arthritis, psoriasis, inflammatory bowel disease, sarcoidosis, T-cell leukemias or transplant rejection.
The other embodiment of the present invention relates to the pharmaceutical formulation comprising Cefazolin as the active ingredient and the pharmaceutically acceptable carriers and/or excipients for the prevention or treatment of the diseases such as rheumatoid arthritis, psoriasis, inflammatory bowel disease, sarcoidosis, T-cell leukemias or transplant rejection.
The pharmaceutical formulation - except for the active ingredient - may contain traditional pharmaceutically acceptable carriers and/or excipients which are inert and do not interact with the active ingredient.
The pharmaceutical formulation - except for the active ingredient - may also contain traditional pharmaceutically acceptable carriers and/or excipients which are inert and do not interact with the active ingredient.
The pharmaceutical composition may be formulated in any pharmaceutical form suitable for the systemic administration, for example oral administration, such as tablets and capsules, starch capsules, coated tablets or enteral tablets; as powders or granules; as a solution, a suspension or an emulsion. Tablets and capsules for oral administration may contain excipients routinely used in pharmaceutical practice, such as binders, diluents, disintegrants or lubricants. The tablets may be coated by any method known in the art. Liquid pharmaceutical compositions for oral administration may be manufactured as, for example, aqueous or oily suspensions, solutions, emulsions, syrups and elixirs or they may be produced as dry substances for the preparation of solutions or suspensions ex tempore with water or other suitable diluent. Liquid pharmaceutical preparations may contain excipients routinely used in pharmaceutical practice, such as dispersing and emulsifying agents, non-aqueous carriers (they may comprise eatable oils) or preservatives. The selection and amount of the excipients depends on the pharmaceutical dosage form and the route of drug administration. The pharmaceutical composition may be formulated into any suitable pharmaceutical form, by any method known in the art, using any pharmaceutically acceptable carriers, diluents, fillers and other excipients.
On account of very poor absorption when Cefazolin is administered orally, parenteral, i.e. intra-venous or intra-muscular route of administration is preferred The pharmaceutical preparation suitable for parenteral administration, for example for intramuscular, subcutaneous or intravenous administration, may be used as a ready suspension, lyophilizate, suspension ex tempore or concentrate for intravenous infusions. These preparations may be formulated as unit dosage forms in ampoules, initially filled syringes, low capacity infusions, or in multi-dosage containers consisting of preservatives and carriers, diluents, stabilizers and/or dispersing agents. The carriers suitable for the intravenous administration of the pharmaceutical preparation comprise, for example, sterile aqueous solutions, such as the solution of physiological salt, the solutions of carbohydrates, for example, glucose, mannitol, dextrose, lactose and aqueous buffer solutions, for example, phosphate buffers. The pharmaceutical composition may also contain other excipients routinely used to maintain isoosmoticity, antioxidants, preservatives and others. Alternatively, the active ingredient may be used as the powder obtained due to isolation of a solid compound under septic conditions or by lyophilisation from the solvent, for the preparation of suspensions ex tempore in a suitable diluent, for example, sterile water deprived of pyrogenic substances. The substance for intra- venous infusions prepared analogously as for the intra-venous injections, can be diluted with glucose or Ringer solution or with aqueous 0,9% sodium chloride and as such can be delivered into the body through a vain.
The biological activity of Cefazolin, inhibiting IL-15 and IL-2-induced cells' response has been proved during in vitro studies. The results also demonstrated inhibition of cell proliferation as well as the synthesis of TNF-a and IL-17, induced by theses two cytokines. When this effectiveness is proved in vivo studies in animal model of the diseases, Cefazolin will have the potential to be used in the prevention and treatment of IL-15 overproduction related diseases and inflammations, such as rheumatoid arthritis, psoriasis, inflammatory bowel disease, sarcoidosis, T-cell leukemias or transplant rejection.
Biological studies In vitro studies
All experiments were carried out on peripheral blood mononuclear cells (PBMC) from healthy donors. The cells were isolated under the standard protocol regarding density gradient centrifugation. According to the general protocol, 6 ml of blood was layered on 3 ml of Lymphoprep (Axis-shield, Norway), then centrifuged at 800 x g for 15 min. The layer of PBMC was collected, the cells were washed twice with phosphate buffered saline (PBS) (BIOMED- LUBLIN, Poland) and suspended in medium RPMI1640 (Gibco, Great Britain), containing 10 mM HEPES (Sigma, USA), 10% fetal calf serum (BIOMED-LUBLIN, Poland) and antibiotic (streptomycin sulfate, penicilline G sodium, amphotericin B, PAA, Austria).
Cefazolin was initially selected at the screening stage, on the basis of the three dimensional structure of the model of [IL-15RaPy - IL-15] protein complex. Cefazolin was used as sodium salt, commercially available as lyophilizate for preparation of suspensions for the injections (Biofazolin, by ZF Polpharma SA)..
The study regarding the assessment of Cefazolin efficacy was performed.
In all the in vitro experiments non stimulated PBMC were used as the control. The inhibition efficacy was assessed in relation to the cells stimulated with IL-15 or IL-2 . Example 1
Effect of Cefazolin on interleukin 15 or interleukin 2-induced PBMC proliferation.
PBMC proliferation was evaluated using commercially available BrdU Cell Proliferation Assay (Calbiochem, Merck, Germany). PBMC were seeded in a 96-well plate (25x103 cells in 200 μΙ of the culture medium/well). The next day the cells were treated with the tested compound at final concentrations: 20 μΜ, 100 μΜ and 300 μΜ. After 30 min. incubation with Cefazolin, cells were stimulated with IL- 5 at the final concentration 5 ng/ml or IL-2 at the final concentration 5 ng/ml. Cells were incubated for 4 days, then for last 24 h bromodeoxyuridine (BrdU) was added to the culture medium at the concentration recommended by the manufacturer. After the completion of the incubation, cells were centrifuged (10 min., 160xg) and fixed. Further experimental steps were performed according to the manufacturer's protocol.
The results of the effect of Cefazolin at different concentrations on IL-15 or IL-2-induced
PBMC proliferation are presented in Fig. 1 and Fig. 2 respectively. The results are expressed as the percentage change of proliferating cells with regard to the control cells stimulated with IL-15 or IL-2. Example 2
Effect of Cefazolin on IL-15 or IL-2-induced TNF-a synthesis in PBMC
The evaluation of TNF-a synthesis in IL-15-stimulated PBMC was performed using the ELISA method and commercially available tests (R&D, USA). The experiment was carried out strictly according to the manufacturer's recommendations.
The PBMC were seeded in a 24-well plate (2x106 cells in 1 ml of the culture medium/well), treated with the tested compound and after 30 min stimulated with IL-15 (5 ng/ml) or IL-2 (5 ng/ml). After 48 h incubation, the culture medium was collected from each well to determine the TNF-a concentration, cells were harvested and lysed, and the concentration of total protein was measured in cell lysates. The obtained values of TNF-a concentration were calculated for 1 mg of protein. The results are expressed as the percentage change of the concentration of TNF-a synthesized in PBMC with regard to the control cells stimulated with II- 15.
The effect of Cefazolin at different concentrations (20 μΜ, 50 μΜ, 100 μΜ, 200 μΜ, 400 μΜ, 600 μΜ, 1000 μΜ) on IL-15-induced TNF-α synthesis in PBMC is depicted in Fig. 3. The effect of Cefazolin at 300 μΜ concentration on IL-2-induced TNF-a synthesis in PBMC is depicted in Fig. 4.
Cefazolin significantly reduces IL-15 or IL-2-induced TNF-a synthesis in PBMC in comparison with IL-15 and IL-2 stimulated control cells. Example 3
Effect of Cefazolin on IL-15 or IL-2-induced IL-17 synthesis in PBMC
The evaluation of IL-17 synthesis in IL-15 or IL-2-stimulated PBMC was performed using the ELISA method and commercially available tests (R&D, USA). The experiment was carried out strictly according to the manufacturer's recommendations.
PBMC were seeded in a 24-well plate (2x106 cells in 1 ml of the culture medium/well), treated with the inhibitor, after 30 min. stimulated with IL-15 (5 ng/ml) or IL-2 (5 ng/ml). After 48 h incubation, the culture medium was collected from each well to determine the IL-17 concentration, cells were harvested and lysed, and the concentration of total protein was measured in cell lysates. The obtained values of IL-17 concentration were calculated for 1 mg of protein. The results are expressed as the percentage change of the concentration of IL-17 synthesized in PBMC with regard to the control cells stimulated with 11-15 or IL-2.
The effect of Cefazolin at different concentrations (20 μΜ, 50 μΜ, 100 μΜ, 200 μΜ, 300 μΜ, 400 μΜ, 600 μΜ and 1000 μΜ) on IL-15-induced IL-17 synthesis in PBMC is depicted in Fig. 5. The effect of Cefazolin at 300 μΜ concentration on IL-2-induced lL-17 synthesis in PBMC is depicted in Fig. 6.
Cefazolin significantly reduces IL-15-induced IL-17 synthesis in PBMC in comparison with IL-15 stimulated control cells. The increase of Cefazolin concentration from 200 μΜ to 1000 μΜ does not affect IL-17 synthesis to any great extend. Strong inhibitory effect of Cefazolin on IL-17 synthesis is observed in cells stimulated with IL-2.
Discussion 1. Initials studies based on the assessment of Cefazolin cytotoxicity and efficacy of inhibition of IL-15 or I L-2-induced cell proliferation.
Increased cell proliferation is one of the characteristic responses to IL-15 or IL-2 stimulation. Selective blocking of the IL-15 specific IL-15Ra receptor as well as shared by IL-15 and IL-2, IL-2RP and IL-2Ry the receptor subunits, results in the inhibition of the biological activity of the said cytokines. As a consequence, IL-15 or I L-2-induced increase of cell proliferation does not occur. The effect of Cefazolin on the proliferation of peripheral blood mononuclear cells (PBMC) isolated from blood of healthy donors was evaluated in the assay using a fluorescent CSFE dye (Molecular Dynamics, Great Britain) and in the bromodeoxyuridine incorporation test (BrdU) (BrdU Cell Proliferation Assay, Calbiochem, USA). The PBMC population consists of several cell types, among others, there are lymphocytes and monocytes. Due to expression of IL-15 and IL-2 receptors on the surface of the membrane, extensive proliferation of these cells occurs in response to interleukin 15 or interleukin 2 stimulation. The inhibition of cell proliferation in the presence of the tested compound can be a visible sign of cells' death caused by the cytotoxic or apoptotic activity of the analyzed compound. The effect of Cefazolin on cells viability was tested measuring the lactate dehydrogenase (LDH) concentration in the culture medium and cultured cells (CytoTox 96 Non- Radioactive Cytotoxicity Assay, Promega, USA). LDH is a cytosolic enzyme which under physiological conditions is not released from cells. However, when cell membrane damage or cell death occurs, LDH enzyme is released into the matrix. Increased LDH activity in the culture medium correlates with the increased number of dead cells resulting from the cytotoxic effect of the tested compound. LDH activity measured in cell lysates is used for the assessment of a number of living cells. The results of the following study has demonstrated biological activity of Cefazolin, which inhibits 11-15 or IL-2-induced PBMC proliferation and does not trigger the apoptotic pathway. The experiments were repeated three time on PBMC from different donors.
2. The assessment of inhibitory effectiveness of Cefazolin on IL-15 and IL-2 -induced TNF-a and I L-17 syntheses.
Peripheral blood mononuclear cells (PBMC) respond to IL-15 or IL-2 stimulation not only by increased proliferation, but also synthesizing many pro-inflammatory cytokines, among others, for example, TNF-a and IL-17. The inhibition of IL-15 and IL-2 biological activity should result in the reduced cell proliferation and decreased syntheses of TNF-a and IL-17.
Cefazolin at different concentrations was used to assess its effect on IL-15 and IL-2- induced other cytokines syntheses. The biological activity of Cefazolin was evaluated measuring the TNF-a and IL-17 concentrations in the culture media collected after the completion of the incubation, using the immunoenzymatic ELISA tests.
The tests results prove the inhibition of the TNF-a and IL- 7 syntheses and they serve as the following evidence of Cefazolin biological activity.

Claims

Claims
7-Aminocephalosporanic acid derivative - Cefazolin, for use as the inhibitor of IL-15 and IL-2 receptors in the prevention and treatment of IL-15 and IL-2 overproduction related diseases.
Cefazolin for use according to claim 1 , wherein the disease is rheumatoid arthritis, psoriasis, inflammatory bowel disease, sarcoidosis, T-cell leukemias or transplant rejection.
Cefazolin for use according to claim 1 , wherein the disease is rheumatoid arthritis.
Cefazolin for use according to claim 1 , wherein Cefazolin is to be administered in a therapeutically effective dose to an individual in need of such a treatment.
Cefazolin for use according to claim 4, wherein Cefazolin is to be administered as a single dosage form.
Use of Cefazolin for manufacturing of the pharmaceutical formulation for the prevention and treatment of the diseases such as rheumatoid arthritis, psoriasis, inflammatory bowel disease, sarcoidosis, T-cell leukemias or transplant rejection.
Pharmaceutical formulation comprising therapeutically effective amount of Cefazolin as the active ingredient and pharmaceutically acceptable carriers and/or excipients for use in the prevention and treatment of the diseases such as rheumatoid arthritis, psoriasis, inflammatory bowel disease, sarcoidosis, T-cell leukemias or transplant rejection.
8. Pharmaceutical formulation according to claim 7, in a form suitable for intra-venous or intra-muscular administration.
PCT/IB2014/001940 2013-09-30 2014-09-29 7-aminocephalosporanic acid derivative as inhibitor of il-15 and il-2 activity WO2015044762A1 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
EP14799527.8A EP3068491B1 (en) 2013-09-30 2014-09-29 7-aminocephalosporanic acid derivative as inhibitor of il-15 and il-2 activity
PL417879A PL235635B1 (en) 2013-09-30 2014-09-29 Applications of the derivative of 7-aminocephalosporanic acid as the L-15 and IL-2 biologic activity inhibitor
PL14799527T PL3068491T3 (en) 2013-09-30 2014-09-29 7-aminocephalosporanic acid derivative as inhibitor of il-15 and il-2 activity
CA2925652A CA2925652A1 (en) 2013-09-30 2014-09-29 7-aminocephalosporanic acid derivative as inhibitor of il-15 and il-2 activity
AU2014326355A AU2014326355B2 (en) 2013-09-30 2014-09-29 7-aminocephalosporanic acid derivative as inhibitor of IL-15 and IL-2 activity
US15/026,125 US20160235762A1 (en) 2013-09-30 2014-09-29 7-aminocephalosporanic acid derivative as inhibitor of il-15 and il-2 activity
DK14799527.8T DK3068491T3 (en) 2013-09-30 2014-09-29 7-Aminocephalosporanoic acid derivative as inhibitor of IL-15 and IL-2 activity
ES14799527T ES2747635T3 (en) 2013-09-30 2014-09-29 7-aminocephalosporic acid derivative as an inhibitor of IL-15 and IL-2 activity
US15/661,586 US11452728B2 (en) 2013-09-30 2017-07-27 7-aminocephalosporanic acid derivative as inhibitor of IL15 and IL-2 activity
AU2019236714A AU2019236714B2 (en) 2013-09-30 2019-09-26 7-aminocephalosporanic acid derivative as inhibitor of IL-15 and IL-2 activity

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
PL40550613A PL405506A1 (en) 2013-09-30 2013-09-30 Application of the 7-aminocephalosporanic acid derivative as inhibitor biological activity of IL-15 and IL-2
PLP.405506 2013-09-30

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US15/026,125 A-371-Of-International US20160235762A1 (en) 2013-09-30 2014-09-29 7-aminocephalosporanic acid derivative as inhibitor of il-15 and il-2 activity
US15/661,586 Division US11452728B2 (en) 2013-09-30 2017-07-27 7-aminocephalosporanic acid derivative as inhibitor of IL15 and IL-2 activity

Publications (1)

Publication Number Publication Date
WO2015044762A1 true WO2015044762A1 (en) 2015-04-02

Family

ID=51903949

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2014/001940 WO2015044762A1 (en) 2013-09-30 2014-09-29 7-aminocephalosporanic acid derivative as inhibitor of il-15 and il-2 activity

Country Status (9)

Country Link
US (2) US20160235762A1 (en)
EP (1) EP3068491B1 (en)
AU (2) AU2014326355B2 (en)
CA (1) CA2925652A1 (en)
DK (1) DK3068491T3 (en)
ES (1) ES2747635T3 (en)
PL (3) PL405506A1 (en)
PT (1) PT3068491T (en)
WO (1) WO2015044762A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3124016A1 (en) 2015-07-31 2017-02-01 Warszawski Uniwersytet Medyczny Antipsoriatic emulsion composition comprising cefazolin

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11278505B2 (en) 2017-04-24 2022-03-22 University Of Massachusetts Diagnosis and treatment of vitiligo

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4891370A (en) 1981-12-14 1990-01-02 Merck & Co., Inc. Cephalosporin derivatives as anti-inflammatory agents
WO2006029578A1 (en) 2004-09-17 2006-03-23 Centro De Ingenieria Genetica Y Biotecnologia Interleukin-15 antagonist peptide
JP2007131535A (en) * 2003-12-26 2007-05-31 Tokai Univ Agent for suppressing formation of modified protein
WO2007065167A1 (en) * 2005-12-02 2007-06-07 The Johns Hopkins University Use of high-dose oxazaphosphorine drugs for treating immune disorders
WO2010037351A1 (en) 2008-09-30 2010-04-08 Centro De Ingenieria Genetica Y Biotecnologia Peptide antagonist of interleukin-15 activity

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1247057A (en) * 1999-06-07 2000-03-15 李桂华 Process for preparing oral cleaner
EP1531828A4 (en) * 2002-05-23 2005-11-02 Activbiotics Inc Methods of treating bacterial infections and diseases associated therewith

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4891370A (en) 1981-12-14 1990-01-02 Merck & Co., Inc. Cephalosporin derivatives as anti-inflammatory agents
JP2007131535A (en) * 2003-12-26 2007-05-31 Tokai Univ Agent for suppressing formation of modified protein
WO2006029578A1 (en) 2004-09-17 2006-03-23 Centro De Ingenieria Genetica Y Biotecnologia Interleukin-15 antagonist peptide
WO2007065167A1 (en) * 2005-12-02 2007-06-07 The Johns Hopkins University Use of high-dose oxazaphosphorine drugs for treating immune disorders
WO2010037351A1 (en) 2008-09-30 2010-04-08 Centro De Ingenieria Genetica Y Biotecnologia Peptide antagonist of interleukin-15 activity

Non-Patent Citations (38)

* Cited by examiner, † Cited by third party
Title
AGOSTINI C.T.L. ET AL., J. IMMUNOL., vol. 157, 1996, pages 910 - 8
ANGIOLLLO A.L. ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 233, 1997, pages 231 - 7
BAAN C.C. ET AL., TRANSPLANT PROC., vol. 31, 1999, pages 2726 - 8
BASLUND B. ET AL., ARTHRITIS RHEUM, vol. 52, 2005, pages 2686 - 92
BUDAGIAN V. ET AL., CYTOKINE GROWTH FACTOR REV., vol. 17, 2006, pages 259 - 80
BUDAGIAN V. ET AL., J. BIOL. CHEM., 2004
CHIODINI R J ET AL: "IN VITRO ANTIMICROBIAL SUSCEPTIBILITY OF A MYCOBACTERIUM SP. ISOLATED FROM PATIENTS WITH CROHN'S DISEASE", ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, AMERICAN SOCIETY FOR MICROBIOLOGY, US, vol. 26, no. 6, 1 December 1984 (1984-12-01), pages 930 - 932, XP000987182, ISSN: 0066-4804 *
DEEPA REDDY ET AL: "Relapses of Inflammatory Bowel Disease During Pregnancy: In-Hospital Management and Birth Outcomes", THE AMERICAN JOURNAL OF GASTROENTEROLOGY, vol. 103, no. 5, 1 May 2008 (2008-05-01), pages 1203 - 1209, XP055159514, ISSN: 0002-9270, DOI: 10.1111/j.1572-0241.2007.01756.x *
DOBBELING U ET AL., BLOOD, vol. 92, 1998, pages 252 - 8
FERRARI-LACRAZ S. ET AL., J. IMMUNOL., vol. 173, 2004, pages 5818 - 26
FERRARI-LACRAZ S. ET AL., TRANSPLANTATION, vol. 82, 2006, pages 1510 - 7
KIM Y.S. ET AL., J. IMMUNOL., vol. 160, 1998, pages 5742 - 8
KIM Y.S., J. IMMUNOL., vol. 160, 1998, pages 5742 - 8
KIRMAN I; NIELSEN O.H., AM. J. GASTROENTEROL, vol. 91, 1996, pages 1789 - 1794
KUNIYASU H. ET AL., PATHOBIOLOGY, vol. 69, 2001, pages 86 - 95
LEWIS E.C ET AL., CYTOKINE, vol. 34, 2006, pages 106 - 13
LIEW F.Y.; MCINNES I.B., ANN. RHEUM. DIS., vol. 61, no. 2, 2002, pages II100 - 2
MCINNES I.B. ET AL., IMMUNOL TODAY, vol. 19, 1998, pages 75 - 9
MCINNES I.B. ET AL., NAT. MED., vol. 2, 1996, pages 175 - 82
MCLNNES I.B ET AL., NAT. MED., vol. 3, pages 189 - 95
MORRIS J.C., PROC. NATL.. ACAD. SCI. USA, vol. 103, 2001, pages 401 - 6
O. ONCUL ET AL: "Serum Interleukin 2 Levels in Patients with Rheumatoid Arthritis and Correlation with Insulin Sensitivity", JOURNAL OF INTERNATIONAL MEDICAL RESEARCH, vol. 30, no. 4, 1 August 2002 (2002-08-01), pages 386 - 390, XP055159734, ISSN: 0300-0605, DOI: 10.1177/147323000203000404 *
RUCHATZ H. ET AL., J IMMUNOL., vol. 160, 1998, pages 5664 - 60
RUCHATZ H., J IMMUNOL., vol. 160, 1998, pages 5664 - 60
SAKAI T. ET AL., GASTROENTEROLOGY, vol. 114, 1998, pages 1237 - 1243
SHI R. ET AL., TRANSPL. IMMUNOL., vol. 12, 2004, pages 103 - 8
SMITH X.G ET AL., J. IMMUNOL., vol. 165, 2000, pages 3444 - 50
SMITH X.S. ET AL., J. IMMUNOL., vol. 165, 2000, pages 3444 - 50
SOMEKH E ET AL: "IN-VITRO EFFECTS OF CIPROFLOXACIN AND PEFLOXACIN ON GROWTH OF NORMAL HUMAN HEMATOPOIETIC PROGENITOR CELLS AND ON LEUKEMIC CELL LINES", JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS, AMERICAN SOCIETY FOR PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS, US, vol. 248, no. 1, 1 January 1989 (1989-01-01), pages 415 - 418, XP009120468, ISSN: 0022-3565 *
TINUBU S.A. ET AL., J. IMMUNOL., vol. 153, 1994, pages 4330 - 8
TREIBER-HELD S; STEWART DM; BARRACLOUGH HA; KURMAN CC; NELSON DL, CLIN IMMUNOL IMMUNOPATHOL., vol. 80, no. 1, July 1996 (1996-07-01), pages 67 - 75
USHIO H. ET AL., LETTERS IN DRUG DESIGN DISCOVERY, vol. 5, 2008, pages 292 - 296
VILLA M L ET AL: "Interference of cephalosporins with immune response: Effects of cefonicid on human T-helper cells", INTERNATIONAL JOURNAL OF IMMUNOPHARMACOLOGY, ELMSFORD,NY, US, vol. 13, no. 8, 1 January 1991 (1991-01-01), pages 1099 - 1107, XP025491690, ISSN: 0192-0561, [retrieved on 19910101], DOI: 10.1016/0192-0561(91)90161-Y *
VILLADSEN L.S ET AL., J. CLIN. INVEST., vol. 112, 2003, pages 1571 - 80
VILLADSEN L.S. ET AL., J. CLIN. INVEST., vol. 112, 2003, pages 1571 - 80
WALDMANN TA., J CLIN IMMUNOL., vol. 27, no. 1, January 2007 (2007-01-01), pages 1 - 18
WEI X ET AL., J. IMMUNOL., vol. 167, 2001, pages 277 - 82
ZHENG X.X. ET AL., TRANSPLANTATION, vol. 81, 2006, pages 109 - 16

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3124016A1 (en) 2015-07-31 2017-02-01 Warszawski Uniwersytet Medyczny Antipsoriatic emulsion composition comprising cefazolin

Also Published As

Publication number Publication date
US20170319593A1 (en) 2017-11-09
ES2747635T3 (en) 2020-03-11
PL235635B1 (en) 2020-09-21
AU2014326355B2 (en) 2019-06-27
EP3068491A1 (en) 2016-09-21
US20160235762A1 (en) 2016-08-18
AU2014326355A1 (en) 2016-04-28
PL405506A1 (en) 2015-04-13
PL417879A1 (en) 2017-02-13
PT3068491T (en) 2019-10-24
AU2019236714B2 (en) 2020-10-15
AU2019236714A1 (en) 2019-10-17
US11452728B2 (en) 2022-09-27
DK3068491T3 (en) 2019-10-14
PL3068491T3 (en) 2019-12-31
CA2925652A1 (en) 2015-04-02
EP3068491B1 (en) 2019-07-03

Similar Documents

Publication Publication Date Title
Furumoto et al. The arrival of JAK inhibitors: advancing the treatment of immune and hematologic disorders
Tanaka Current concepts in the management of rheumatoid arthritis
Maeshima et al. The JAK inhibitor tofacitinib regulates synovitis through inhibition of interferon‐γ and interleukin‐17 production by human CD4+ T cells
Nakano et al. Dopamine induces IL-6–dependent IL-17 production via D1-like receptor on CD4 naive T cells and D1-like receptor antagonist SCH-23390 inhibits cartilage destruction in a human rheumatoid arthritis/SCID mouse chimera model
JP6263468B2 (en) Combination of MEK inhibitor and selective inhibitor of Aurora A kinase
Netea et al. Pharmacologic inhibitors of tumor necrosis factor production exert differential effects in lethal endotoxemia and in infection with live microorganisms in mice
Adami et al. The role of interleukin-17A in psoriatic disease
Ali et al. Canonical stimulation of the NLRP3 inflammasome by fungal antigens links innate and adaptive B-lymphocyte responses by modulating IL-1β and IgM production
AU2019236714B2 (en) 7-aminocephalosporanic acid derivative as inhibitor of IL-15 and IL-2 activity
US9301961B2 (en) Autoimmune and inflammatory disorder therapy
EP1450775B1 (en) C-kit inhibitors for treating bacterial infections
JPH04506215A (en) Monokine activity interference
JP6093760B2 (en) Leukotoxin E / D as anti-inflammatory and bactericidal agent
US20170198011A1 (en) Composition for treating sepsis or septic shock comprising the peptide originated from the smad6
JP2009529529A (en) Use of VX-702 to treat rheumatoid arthritis
WO2014191822A1 (en) BENZOIC ACID DERIVATIVES AS IL-15Rα RECEPTOR INHIBITORS
Matzer et al. A synthetic, non-peptide CXCR2 antagonist blocks MIP-2-induced neutrophil migration in mice
US9855225B2 (en) Cannabinoid receptor treatments
US20070117833A1 (en) Pharmaceutical compositions
US20120039867A1 (en) Immune System Function in Conditions Characterized by Elevated Double Strand Breaks
WO2014191823A1 (en) Amine derivatives as il-15 activity inhibitors
WO2015044761A1 (en) Diaminodecanedioic acid derivative as the inhibitor of il-15 and il-2 activity
Tanaka et al. JAK and SYK: emerging their relevance to the treatment of inflammatory diseases
Hartmann et al. Blockade of TNF does not alter oxygen burst and phagocytosis of human neutrophils in patients with rheumatoid arthritis
Tanaka et al. Cytokine-mediated signals as targets for treatment of rheumatoid arthritis: a JAK inhibitor in vitro and in vivo

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14799527

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2925652

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 15026125

Country of ref document: US

Ref document number: 417876

Country of ref document: PL

ENP Entry into the national phase

Ref document number: 2014326355

Country of ref document: AU

Date of ref document: 20140929

Kind code of ref document: A

REEP Request for entry into the european phase

Ref document number: 2014799527

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2014799527

Country of ref document: EP